throbber
V O L U M E 2 9 䡠 N U M B E R 1 8 䡠 J U N E 2 0 2 0 1 1
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`O R I G I N A L R E P O R T
`
`Phase I Study of Oral Azacitidine in Myelodysplastic
`Syndromes, Chronic Myelomonocytic Leukemia, and Acute
`Myeloid Leukemia
`Guillermo Garcia-Manero, Steven D. Gore, Christopher Cogle, Renee Ward, Tao Shi, Kyle J. MacBeth,
`Eric Laille, Heidi Giordano, Sarah Sakoian, Elias Jabbour, Hagop Kantarjian, and Barry Skikne
`
`A
`
`B
`
`S
`
`T
`
`R
`
`A
`
`C
`
`T
`
`Purpose
`To determine the maximum-tolerated dose (MTD), safety, pharmacokinetic and pharmacody-
`namic profiles, and clinical activity of an oral formulation of azacitidine in patients with
`myelodysplastic syndromes (MDSs), chronic myelomonocytic leukemia (CMML), or acute
`myeloid leukemia (AML).
`
`Patients and Methods
`Patients received 1 cycle of subcutaneous (SC) azacitidine (75 mg/m2) on the first 7 days of cycle
`1, followed by oral azacitidine daily (120 to 600 mg) on the first 7 days of each additional 28-day
`cycle. Pharmacokinetic and pharmacodynamic profiles were evaluated during cycles 1 and 2.
`Adverse events and hematologic responses were recorded. Cross-over to SC azacitidine was
`permitted for nonresponders who received ⱖ 6 cycles of oral azacitidine.
`
`Results
`Overall, 41 patients received SC and oral azacitidine (MDSs, n ⫽ 29; CMML, n ⫽ 4; AML, n ⫽ 8).
`Dose-limiting toxicity (grade 3/4 diarrhea) occurred at the 600-mg dose and MTD was 480 mg.
`Most common grade 3/4 adverse events were diarrhea (12.2%), nausea (7.3%), vomiting (7.3%),
`febrile neutropenia (19.5%), and fatigue (9.8%). Azacitidine exposure increased with escalating
`oral doses. Mean relative oral bioavailability ranged from 6.3% to 20%. Oral and SC azacitidine
`decreased DNA methylation in blood, with maximum effect at day 15 of each cycle. Hematologic
`responses occurred in patients with MDSs and CMML. Overall response rate (ie, complete
`remission, hematologic improvement, or RBC or platelet transfusion independence) was 35% in
`previously treated patients and 73% in previously untreated patients.
`
`Conclusion
`Oral azacitidine was bioavailable and demonstrated biologic and clinical activity in patients with
`MDSs and CMML.
`
`J Clin Oncol 29:2521-2527. © 2011 by American Society of Clinical Oncology
`
`INTRODUCTION
`
`Azacitidine is a cytidine nucleoside analog with a
`mechanism of action that involves incorporation
`into DNA and RNA.1,2 Data suggest that patients
`must be exposed to azacitidine over several treat-
`ment cycles for optimal therapeutic effect.3 The
`requirement for chronic exposure can be ex-
`plained by drug pharmacokinetics, as azacitidine
`has a short plasma half-life, and by mechanism of
`action, as induction of DNA hypomethylation
`through incorporation into DNA is cell-cycle
`dependent (S-phase restricted) and DNA rem-
`ethylation is observed by the end of each treat-
`ment cycle.4
`
`A treatment regimen facilitating chronic ad-
`ministration may help achieve optimal efficacy
`outcomes. An oral azacitidine formulation would
`improve convenience of administration and ex-
`pand the possibilities of exploring novel mainte-
`nance schedules, targeting different malignancies,
`and testing multiple combinations. A phase 0 trial
`demonstrated that a single oral azacitidine dose
`resulted in detectable levels in the blood.5
`This phase I study sought to identify the
`maximum-tolerateddose(MTD),dose-limitingtoxic-
`ities (DLTs), safety, pharmacokinetic and pharmaco-
`dynamicprofiles,andclinicalactivityoforalazacitidine
`in patients with myelodysplastic syndromes (MDSs),
`chronic myelomonocytic leukemia (CMML), or acute
`myeloid leukemia (AML).
`
`© 2011 by American Society of Clinical Oncology
`
`2521
`
`Guillermo Garcia-Manero, Elias Jabbour,
`Hagop Kantarjian, University of Texas
`MD Anderson Cancer Center, Houston,
`TX; Steven D. Gore, Sarah Sakoian, The
`Sidney Kimmel Comprehensive Cancer
`Center at Johns Hopkins, Baltimore,
`MD; Christopher Cogle, University of
`Florida, Gainesville, FL; Renee Ward,
`Tao Shi, Kyle J. MacBeth, Eric Laille,
`Heidi Giordano, Barry Skikne, Celgene,
`Summit, NJ; Barry Skikne, University of
`Kansas Medical Center, Kansas City, KS.
`
`Submitted December 21, 2010;
`accepted March 22, 2011; published
`online ahead of print at www.jco.org on
`May 16, 2011.
`
`Supported by Celgene; editorial support
`in the preparation of this manuscript
`from Excerpta Medica, sponsored by
`Celgene.
`
`Authors’ disclosures of potential con-
`flicts of interest and author contribu-
`tions are found at the end of this
`article.
`
`Clinical Trials repository link available on
`JCO.org.
`
`Corresponding author: Guillermo Garcia-
`Manero, MD, University of Texas, MD
`Anderson Cancer Center, Box 428,
`1515 Holcombe Blvd, Houston, TX
`77025; e-mail: ggarciam@mdanderson
`.org.
`
`© 2011 by American Society of Clinical
`Oncology
`
`0732-183X/11/2918-2521/$20.00
`
`DOI: 10.1200/JCO.2010.34.4226
`
`CELGENE 2084
`APOTEX v. CELGENE
`IPR2023-00512
`
`Downloaded from ascopubs.org by 72.165.200.210 on March 25, 2021 from 072.165.200.210
`Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
`
`

`

`Garcia-Manero et al
`
`PATIENTS AND METHODS
`
`The trial was approved by the relevant institutional review boards and ethics
`committees. All patients gave written informed consent.
`
`Patients
`Eligible patients were ⱖ 18 years, had an Eastern Cooperative Oncology
`Group performance status score of 0 to 2, and a diagnosis of MDSs, CMML, or
`AML according to WHO classification.6,7 For patients with AML, eligibility
`was limited to those for whom standard curative measures did not exist or were
`no longer effective. Exclusion criteria included a diagnosis of acute promyelo-
`cytic leukemia, previous treatment with hypomethylating agents within 4
`weeks before cycle 1, and anticancer therapy within 21 days before the first dose
`
`Table 1. Baseline Patient Characteristics (N ⫽ 41)
`
`Parameter
`
`No. of Patients
`
`%
`
`Median age, years
`Range
`Sex
`Male
`Female
`MDSs (WHO classification)
`RA/RARS/RCMD
`RAEB-1
`RAEB-2
`MDSs-U
`CMML
`AML
`De novo
`Transformed from MDSs
`IPSS (MDSs patients)ⴱ
`Low risk
`Intermediate 1 risk
`Intermediate 2 risk
`High risk
`Not available†
`Hematology
`Median hemoglobin, g/dL
`Range
`Median white blood cell count ⫻109/L
`Range
`Median absolute neutrophil count ⫻109/L
`Range
`Median platelet count ⫻109/L
`Range
`Cytogenetics‡
`Normal chromosomal karyotype
`1 chromosomal abnormality
`2 chromosomal abnormalities
`ⱖ 3 chromosomal abnormalities
`Prior treatment with hypomethylating agent
`MDSs
`CMML
`AML
`
`70
`31-91
`
`32
`9
`29
`11
`12
`5
`1
`4
`8
`4
`4
`
`2
`12
`13
`1
`1
`
`9.3
`6.9-15.1
`2.4
`0.4-30.2
`0.8
`0.0-21.7
`54.0
`3.0-262.0
`
`17
`9
`3
`6
`16
`13
`0
`3
`
`78
`22
`71
`27
`29
`12
`2
`10
`20
`10
`10
`
`7
`41
`45
`3
`3
`
`49
`26
`9
`17
`39
`32
`0
`7
`
`Abbreviations: AML, acute myeloid leukemia; CMML, chronic myelomono-
`cytic leukemia; IPSS, International Prognostic Scoring System; MDSs, myelo-
`dysplastic syndromes; MDSs-U, MDSs unclassified; RA, refractory anemia;
`RAEB, RA with excess blasts; RARS, RA with ringed sideroblasts; RCMD,
`refractory cytopenias with multilineage dysplasia.
`ⴱIPSS score11 was available for 28 patients with MDSs.
`†Patient had a bone marrow transplantation and therefore IPSS risk was not
`considered applicable.
`‡Cytogenetic data were available for 35 patients.
`
`of study drug, or less than full recovery from any significant toxic effects of
`prior treatments.
`
`Study Design and Therapy
`This open-label, phase I, dose-escalation trial was performed in four
`participating institutions and evaluated multiple cycles of oral azacitidine
`administered daily for the first 7 days of a 28-day cycle. The objectives were to
`determine the MTD, DLTs, and the safety profile of oral azacitidine. Pharma-
`cokinetic and pharmacodynamic profiles of oral and subcutaneous (SC) azaci-
`tidine, administered on the same 7-day schedule, were also compared. A
`secondary objective was to assess the clinical activity of oral azacitidine.
`During cycle 1, patients received azacitidine 75 mg/m2 daily SC for 7 days
`of a 28-day cycle. During cycle 2 and beyond, patients received oral azacitidine
`under fasting conditions (ie, no food for 2 hours before and after dosing). The
`dose of oral azacitidine was escalated following a standard phase I 3 ⫹ 3 design.
`The starting dose was 120 mg and doses were escalated in 60 mg increments up
`to a dose of 360 mg, followed by 120 mg increments until the MTD was
`reached. Intrapatient dose escalation was permitted if the dose level to which
`the patient was escalated was associated with a DLT rate of ⱕ 33%. Treatment
`continued until disease progression, lack of activity, unacceptable toxicity, or
`patient preference.
`The MTD was defined as the highest dose at which no more than 33% of
`patients experienced a DLT. DLT was defined as: grade ⱖ 3 nausea, diarrhea,
`or vomiting despite adequate/maximal medical intervention; grade ⱖ 3 clini-
`cally significant nonhematologic toxicity unrelated to underlying disease or
`intercurrent illness; failure to recover to an absolute neutrophil count (ANC)
`of higher than 500/␮L and/or platelet count of higher than 25,000/␮L with
`hypocellular bone marrow (⬍ 5%) 42 days after starting oral azacitidine
`(patients with a baseline ANC of ⱕ 500/␮L and/or platelet count of
`ⱕ 25,000/␮L were not evaluable for neutrophil or platelet toxicity); any
`treatment-related effect resulting in missing ⱖ 3 oral azacitidine doses in the
`7-day treatment period; or any treatment-related nonhematologic toxicity
`delaying initiation of the second oral azacitidine cycle by longer than 14 days.
`Only DLTs that occurred during the first oral azacitidine cycle were considered
`in determining the MTD. Adverse events were graded using the National
`Cancer Institute Common Toxicity Criteria for Adverse Events version 3.0.
`
`Table 2. Incidence of Adverse Events According to Severity in ⱖ 20% of
`Patients Treated With Oral Azacitidine (n ⫽ 41)
`
`CTCAE Grade
`
`System Organ Class
`Preferred Term
`(MeDRA 10.1)
`
`Diarrhea
`Nausea
`Constipation
`Vomiting
`Abdominal pain
`Headache
`Fatigue
`Peripheral edema
`Fever
`Cough
`Contusion
`Dizziness
`Febrile neutropenia
`
`1
`
`2
`
`3
`
`4
`
`Total
`
`No. % No. % No. % No. % No. %
`
`10 24.4 12 29.3
`8 19.5 10 24.4
`9 22.0
`7 17.1
`4
`9.8
`6 14.6
`6 14.6
`4
`9.8
`7 17.1
`5 12.2
`6 14.6
`2
`4.9
`11 26.8
`1
`2.4
`6 14.6
`2
`4.9
`7 17.1
`1
`2.4
`9 22.0
`0
`5 12.2
`3
`0
`0
`
`7.3
`
`4
`3
`0
`3
`0
`1
`4
`0
`2
`2
`0
`0
`8
`
`9.8
`7.3
`
`7.3
`
`2.4
`9.8
`
`4.9
`4.9
`
`19.5
`
`1
`0
`0
`0
`0
`0
`0
`0
`0
`0
`0
`0
`0
`
`2.4 27 65.9
`21 51.2
`16 39.0
`13 31.7
`10 24.4
`13 31.7
`12 29.3
`12 29.3
`10 24.4
`10 24.4
`9 22.0
`8 19.5
`8 19.5
`
`NOTE. This Table includes all adverse events which started during any dosing
`cycle at which oral azacitidine was administered. Percentages are based on
`the number of patients who received at least one dose of oral azacitidine.
`Multiple reports of the same preferred term from a patient are counted only
`once, using the maximum CTCAE grade.
`Abbreviations: CTCAE, National Cancer Institute Common Toxicity Criteria
`for Adverse Events; MedDRA, Medical Dictionary for Regulatory Activities.
`
`2522
`
`© 2011 by American Society of Clinical Oncology
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`Downloaded from ascopubs.org by 72.165.200.210 on March 25, 2021 from 072.165.200.210
`Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
`
`

`

`Phase I Study of Oral AZA in MDSs, CMML, and AML
`
`Pharmacokinetic Analysis
`Plasma and urine pharmacokinetic evaluation of azacitidine was per-
`formed on days 1 and 7 in cycles 1 and 2. Samples were collected up to 8 hours
`after administration and analyzed using a validated high-performance liquid
`chromatography/tandem mass spectrometric method. Parameters calculated
`using noncompartmental method, included maximum observed plasma con-
`centration (Cmax), time of maximum observed plasma concentration (Tmax),
`area under the plasma concentration–time curve from zero to infinity
`(AUCinf), apparent total clearance (CL/F), relative oral bioavailability (F), and
`apparent volume of distribution (Vd/F).
`Pharmacodynamic Analysis
`DNA methylation levels were measured to determine DNA hypomethy-
`lating activity of azacitidine when administered SC or orally. Whole blood was
`collected at baseline and before drug administration on days 3, 8, 15, and 22 of
`cycle 1, and days 1, 3, 8, 15, 22, and 28 of cycle 2. Genomic DNA was purified
`from each whole blood sample using the PAXgene Blood DNA System (Qia-
`gen; Valencia, CA). DNA methylation was analyzed using the Infinium Hu-
`man Methylation27 BeadArray (Illumina; San Diego, CA). In cycle 1, DNA
`methylation data were generated from blood samples of 15 patients. For 10 of
`these patients, data were also generated in cycle 2. A methylation ratio, or beta
`
`value, for each locus per sample was calculated as methylated signal/(methyl-
`ated ⫹ unmethylated signal). Those with detection P ⱕ .05 were considered
`high-quality measures. Samples with more than 25,200 high-quality beta val-
`ues and 26,304 autosomal loci with high-quality beta values in at least half of
`the samples were used for analyses. The low-quality beta values were reim-
`puted using the pamr.knnimpute function from the R package pamr.8 Wil-
`coxon signed-rank tests were performed to identify loci with significant
`methylation differences at each post-treatment time point versus baseline;
`P ⬍ .01 was considered statistically significant. All statistical analyses were
`carried out in R (R Foundation for Statistical Computing, Vienna, Austria,
`http://www.R-project.org).
`Clinical Activity
`Data for clinical activity were evaluated using International Working
`Group (IWG) 2006 criteria, with modifications as described below, for pa-
`tients with MDSs or CMML9 and IWG 2003 criteria for patients with AML.10
`Complete remission (CR), hematologic improvement (HI), and RBC and
`platelet transfusion independence (TI) were evaluated for patients with MDSs
`or CMML. Bone marrow CR (mCR) was also evaluated but not included in the
`overall response rate. RBC transfusion dependence at baseline was defined
`as ⱖ 4 RBC units in the 56 days before cycle 1. Platelet transfusion dependence
`
`SC AZA
`
`Oral AZA
`
`8,000
`
`6,000
`
`4,000
`
`2,000
`
`0
`
`Baseline
`
`Day 3
`
`Day 8
`
`Day 15
`Day 22
`Cycle 1
`
`End
`
`Day 3
`
`End
`
`Day 8
`
`Day 15
`Day 22
`Cycle 2
`
`SC azacitidine
`cycle 1, day 15
`
`Oral azacitidine
`cycle 2, day 15
`
`63
`5,435
`
`1
`1,482
`
`8
`118
`
`No. of Hypermethylated Loci
`
`B
`
`D
`
`SC 75 mg/m2, day 1 (n = 42)
`Oral 480 mg, day 1 (n = 14)
`
`1
`
`2
`
`3
`4
`5
`Time (hours)
`
`6
`
`7
`
`8
`
`SC AZA
`
`Oral AZA
`
`900
`
`800
`
`700
`
`600
`
`500
`
`400
`
`300
`
`200
`
`100
`
`0
`
`0.2
`
`0.1
`
`0.0
`
`-0.1
`
`-0.2
`
`-0.3
`
`-0.4
`
`A
`
`Concentration (ng/mL)
`
`Azacitidine Plasma
`
`C
`
`Change Versus Baseline
`
`Average Methylation
`
`Baseline
`
`Day 3
`
`Day 8
`
`Day 15
`Day 22
`Cycle 1
`
`End
`
`Day 3
`
`End
`
`Day 8
`
`Day 15
`Day 22
`Cycle 2
`
`(B)
`(A) Mean azacitidine (AZA) plasma concentration versus time profiles following single subcutaneous (SC) or oral administration (linear scale).
`Fig 1.
`Pharmacodynamics as measured by plotting the numbers of highly methylated loci (beta ⱖ 0.7; ⫾ 95% CI) for 10 patients with DNA methylation data in cycles 1 and
`2 (gold lines represent individual patients, blue line represents the average). (C) Change in methylation level during treatment with SC or oral AZA for 5,232 loci highly
`methylated at baseline (blue box represents the 25th to 75th percentile, horizontal band represents the median, vertical line with bars represents minimum and
`maximum values). (D) Number of significantly differentially methylated loci on day 15 of cycle 1 (SC azacitidine) and on day 15 of cycle 2 (oral azacitidine). Upward arrows
`denote hypermethylated loci and downward arrows denote hypomethylated loci.
`
`www.jco.org
`
`© 2011 by American Society of Clinical Oncology
`
`2523
`
`Downloaded from ascopubs.org by 72.165.200.210 on March 25, 2021 from 072.165.200.210
`Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
`
`

`

`Garcia-Manero et al
`
`at baseline was defined as ⱖ 2 platelet transfusions in the 56 days before cycle 1
`(modification to IWG 2006 criteria). RBC and platelet TI were defined as no
`transfusions in any 56 consecutive-day period on treatment. Patients who
`achieved ⱖ 50% reduction in platelet transfusion requirement, but not platelet
`TI, in any 56 consecutive-day period on treatment were counted as having
`achieved HI platelet (HI-P; modification to IWG 2006 criteria). Patients RBC
`transfusion dependent at baseline achieving a ⱖ 50% reduction in RBC trans-
`fusion requirement in any 56-consecutive day period and patients not RBC
`transfusion dependent at baseline, but who achieved a 1.5 g/dL increase in
`hemoglobin in any 56-consecutive day period on treatment were considered
`to have achieved HI erythroid (HI-E; modification to IWG 2006 criteria). All
`patients who received ⱖ 1 cycle of oral azacitidine were included in the
`response analysis. The cutoff date for data in this article was August 19, 2010.
`
`RESULTS
`
`Patient Characteristics
`Forty-five patients were treated on a 7-day once-daily schedule.
`Four patients received the first cycle of SC azacitidine only; three
`discontinued due to progressive disease (including one death), and
`one withdrew consent. Baseline characteristics for the remaining 41
`patients who received oral azacitidine are presented in Table 1.11
`Cytogenetic data were available at baseline for 35 of 41 patients treated
`with oral azacitidine; nearly half of the patients had normal karyotype,
`approximately 25% had a single abnormality, and nearly 20% had a
`complex karyotype (ⱖ 3 chromosomal abnormalities). Overall, 16
`(39%) of 41 patients had received prior hypomethylating therapy.
`
`Dose Escalation of Oral Azacitidine
`No DLTs were observed at dose levels up to 480 mg. DLT was
`observed at the 600 mg dose, with two (66.7%) of three patients
`experiencing severe diarrhea, despite adequate medical intervention
`(grade 3 in one patient and grade 4 in the other). Per protocol, the
`MTD was exceeded and the previous dose level of 480 mg was deter-
`mined to be the MTD.
`
`Safety Profile
`Table 2 shows the incidence of AEs (any grade) that oc-
`curred in ⱖ 20% of patients treated with oral azacitidine. The most
`
`frequently observed AEs were gastrointestinal disorders, headache,
`fatigue, and peripheral edema. Other commonly occurring AEs
`included fever, cough, contusion, dizziness, and febrile neutrope-
`nia. Grade 3/4 nausea and grade 3/4 vomiting were each observed
`in 7% of patients. Grade 3 fatigue was observed in 10% of patients.
`Diarrhea occurred at grade 3 severity in 10% of patients and grade
`4 severity in 2%. Grade 3 febrile neutropenia was observed in eight
`patients (20%), with four of those having an ANC of ⱕ 500/␮L
`at baseline.
`Of the 41 patients who received oral azacitidine, 33 terminated
`from the study as of the date of data analysis, with 17 discontinuing
`before completing 6 cycles of oral therapy. Reasons for discontinua-
`tion included disease progression/treatment failure (n ⫽ 10), investi-
`gator decision primarily due to absence of observed benefit/response
`(n ⫽ 15), withdrawal of consent (n ⫽ 4), AEs (n ⫽ 3), and decision to
`pursue hematopoietic stem-cell transplantation (n ⫽ 1). There were
`three deaths within 28 days of last dose of study drug due to multiple
`organ failure (n ⫽ 1), gastrointestinal hemorrhage (n ⫽ 1), and
`pneumonia plus urinary tract infection (n ⫽ 1). No deaths were
`attributed to study drug. Eight patients remained on the study at the
`time of data analysis, having each received between 14 and 32 treat-
`ment cycles.
`
`Pharmacokinetic Characteristics of Azacitidine
`High interpatient variability was noted for all pharmacokinetic
`parameters. Azacitidine was rapidly absorbed after SC (n ⫽ 42) and
`oral (n ⫽ 36) administration, reaching Cmax within 0.5 hours (range,
`0.2 to 1.1 hours) and 1.0 hours (range, 0.3 to 3.6 hours) postdose,
`respectively. Concentration versus time profiles decreased in a pseu-
`dobiphasic manner (Fig 1A). The mean elimination half-life was 1.6 ⫾
`0.7 hours for SC and 0.62 ⫾ 0.25 hours for oral azacitidine. Exposure
`after single oral administration generally increased with dose (Table
`3). For the seven oral dose levels, the mean relative azacitidine oral
`bioavailability (F) ranged from 6.3% to 20%. The MTD had a mean
`relative bioavailability of 13% ⫾ 9%. CL/F exceeded hepatic blood
`flow, indicating extrahepatic metabolism, and Vd/F was greater than
`total body water, suggesting extensive tissue distribution. The amount
`of azacitidine recovered in urine relative to dose was small (⬍ 2%) for
`
`Table 3. Day 1 Plasma Pharmacokinetics Parameters After Single Subcutaneous or Oral Azacitidine Administration
`
`AUCinf
`(ng ⫻ h/mL)
`Tmax (h)
`Cmax (ng/mL)
`CL/F (L/h)
`Mean SD %CV Mean
`SD %CV Mean SD %CV Median Range Mean
`
`No. of
`Patients
`
`F (%)
`
`Vd/F (L)
`SD %CV Mean SD
`
`Relative Oral
`Bioavailability
`
`42
`
`4
`3
`3
`5
`5
`14
`2
`
`1,020 440
`
`43ⴱ
`
`175
`
`128
`
`73ⴱ
`
`650
`
`250
`
`39
`
`0.50
`
`0.2-1.1
`
`410
`
`410 101ⴱ NA
`
`43
`62
`64
`112
`463 221
`282
`88
`311 141
`362 253
`502 100
`
`70
`58
`48
`31
`45
`70
`20
`
`4,100 4,860 118
`2,330 1,890
`81
`598
`258
`43
`1,180
`487
`41
`1,360
`573
`42
`2,140 1,620
`76
`1,220
`244
`20
`
`38
`72
`215
`144
`195
`211
`253
`
`24
`36
`102
`13
`79
`140
`29
`
`64
`50
`47
`9.2
`40
`66
`12
`
`1.48
`1.50
`1.00
`1.48
`1.00
`1.00
`1.50
`
`1.0-2.0 2,930 3,810 130
`1.0-1.5 1,700 1,580
`93
`1.0-1.5
`814
`421
`52
`1.0-2.0 1,090
`626
`57
`0.5-3.6
`947
`251
`27
`0.3-2.5 2,010 1,910
`95
`1.0-2.0 1,580 1,410
`89
`
`8.1
`6.3
`20.0
`11.5
`12.8
`12.8
`14.9
`
`5.6
`2.3
`9.6
`2.6
`2.4
`9.4
`0.8
`
`69
`37
`48
`23
`19
`74†
`5
`
`Dose
`
`Subcutaneous,
`75 mg/m2
`Oral, mg
`120
`180
`240
`300
`360
`480
`600
`
`Abbreviations: AUCinf, area under the plasma concentration–time curve from time zero to infinity; CL/F, apparent total clearance; Cmax, maximum observed plasma
`concentration; F, relative oral bioavailability; NA, not applicable; Tmax, time of maximum observed plasma concentration; Vd/F, apparent volume of distribution.
`ⴱn ⫽ 40.
`†n ⫽ 13.
`
`2524
`
`© 2011 by American Society of Clinical Oncology
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`Downloaded from ascopubs.org by 72.165.200.210 on March 25, 2021 from 072.165.200.210
`Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
`
`

`

`Phase I Study of Oral AZA in MDSs, CMML, and AML
`
`SC and oral administration, suggesting that nonrenal elimination
`is the predominant pathway for clearance. Results after multiple
`doses were similar to those obtained after a single dose for both
`administration routes (data not shown). There was no evidence of
`azacitidine accumulation.
`
`Pharmacodynamics of Azacitidine: Effect on
`DNA Methylation
`DNA methylation was evaluated during cycles 1 and 2 in 10
`patients treated with oral azacitidine. The numbers of highly methyl-
`ated loci were calculated at each time point by averaging across pa-
`tients the number of loci with methylation ratios ⱖ 0.7 (Fig 1B). These
`numbers decreased after SC and oral administration, with maximal
`effects at day 15 of each cycle. The reduction in levels of highly meth-
`ylated loci was not maintained throughout the entire cycle and re-
`turned to near-baseline levels by the end of each cycle. SC azacitidine
`decreased a greater number of loci in comparison to oral azacitidine.
`The changes in methylation level from baseline across patients for the
`5,232 highly methylated loci (average methylation ratio at baseline
`ⱖ 0.7) are represented as box plots (Fig 1C). As with the analysis of
`total numbers of highly methylated loci, the median DNA methyl-
`
`ation of these loci was reduced by 0.115 on day 15 of cycle 1 (SC
`azacitidine) and 0.055 on day 15 of cycle 2 (oral azacitidine), and
`returned to baseline levels at the end of each cycle.
`Differentially methylated loci at each post-treatment time point
`compared with baseline were identified in cycles 1 and 2, with the
`maximum number observed on day 15 of each cycle; 6,981 loci were
`differentially methylated (6,917 hypomethylated) on day 15 of cycle 1
`(SC azacitidine) and 1,609 loci were differentially methylated (1,600
`hypomethylated) on day 15 of cycle 2 (oral azacitidine; P ⬍ .01). In
`total, 1,482 loci were significantly hypomethylated by both SC and oral
`azacitidine (Fig 1D), representing 92.6% of all loci significantly hy-
`pomethylated by oral azacitidine treatment. These data demonstrate
`comparable biologic activity with SC and oral azacitidine, albeit to a
`lesser extent with oral azacitidine.
`
`Clinical Activity of Oral Azacitidine
`The median number of oral azacitidine cycles administered to
`patients with MDSs, CMML, and AML was 6 (range, 1 to 32⫹), 12.5⫹
`(range, 3 to 28⫹), and 4.5 (range, 1 to 15), respectively. Treatment
`duration is summarized in Figure 2. The number of patients from the
`MDSs, CMML, and AML groups who remained on the study at the
`
`MDSs-U
`RCMD
`RAEB-1
`RAEB-1
`RCMD
`RA
`RAEB-2
`RA
`RCMD
`RAEB-1
`RAEB-1
`RAEB-2
`RCMD
`RAEB-1
`RA
`RCMD
`RAEB-2
`RAEB-2
`RAEB-1
`RAEB-1
`RA
`RAEB-1
`RAEB-1
`RCMD
`RCMD
`RAEB-1
`RAEB-1
`RAEB-1
`RAEB-2
`
`CMML
`CMML
`CMML
`CMML
`
`AML
`AML
`AML
`AML
`AML
`AML
`AML
`AML
`
`Fig 2. Treatment duration for the 41 pa-
`tients treated with oral azacitidine (AZA).
`AML, acute myeloid leukemia; CMML,
`chronic myelomonocytic leukemia; MDSs-U,
`myelodysplastic syndromes-unclassified; RA,
`refractory anemia; RCMD, refractory cyto-
`penias with multilineage dysplasia; RAEB,
`RA with excess blasts; SC, subcutaneous.
`
`Initial SC cycle
`Oral cycles, active
`Oral cycles, discontinued
`Crossover SC cycles, discontinued
`
`0
`
`5
`
`10
`
`15
`20
`No. of Cycles Initiated
`
`25
`
`30
`
`35
`
`www.jco.org
`
`© 2011 by American Society of Clinical Oncology
`
`2525
`
`Downloaded from ascopubs.org by 72.165.200.210 on March 25, 2021 from 072.165.200.210
`Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
`
`

`

`Garcia-Manero et al
`
`Table 4. Response in Myelodysplastic Syndromes and Chronic Myelomonocytic Leukemia Patients
`
`Previously Treated Patientsa
`
`First-Line Treatment
`
`Response
`
`Responders
`
`Evaluable Patients
`
`Overall responseb
`CRd
`Any HIe
`HI-E
`HI-N
`HI-P
`
`TI
`
`Red blood cell
`Platelet
`mCRe,f
`
`6
`0
`6
`3
`0
`5
`0
`0
`0
`6
`
`17
`17
`16
`10
`10
`14
`5
`3
`4
`9
`
`%
`
`35
`0
`38
`30
`0
`36
`0
`0
`0
`67
`
`Responders
`
`Evaluable Patients
`
`11
`6
`5
`2
`2
`2
`1
`1
`0
`2
`
`15
`15
`9
`4
`7
`6
`3
`3
`
`6
`
`%
`
`73
`40
`56
`50
`29
`33
`33
`33
`
`33
`
`Duration of
`Response:Range
`(days)
`
`30-483c
`30-152
`56-483c
`56-483c
`82-321c
`58-351c
`76
`76
`NA
`63-422g
`
`NOTE. At any cycle of azacitidine, International Working Group 2006 criteria were used with modifications as described in the Patients and Methods section.
`Abbreviations: CR, complete remission; E, erythroid; HI, hematologic improvement; mCR, bone marrow complete remission; N, neutrophil; NA, not applicable; P,
`platelet; TI, transfusion independence.
`aIncludes erythropoiesis-stimulating agents, chemotherapy, hypomethylating agents, and investigational and/or other agents.
`bOverall response rate does not include patients achieving mCR only.
`cOne or more responses, including that at upper limit of range, are ongoing. Data were censored as of last visit entered into the clinical database.
`dPatients achieving CR were not included in any other categories.
`eOne patient with mCR in the previously treated group also achieved HI (both HI-E and HI-P). Two patients with mCR in the first-line treatment group also achieved
`HI (one patient with HI-P and one patient with both HI-E and HI-N). These patients have been included in both the mCR and HI categories.
`fIn the eight patients who achieved mCR, the response began in cycle 1 of subcutaneous (SC) dosing (n ⫽ 4) or very early in cycle 2 of oral dosing (n ⫽ 4). Therefore,
`the contribution of a single SC azacitidine cycle to the induction of these responses is likely relevant.
`gBone marrow aspirates were not required after 6 cycles of oral azacitidine treatment, therefore follow-up data were not available to confirm upper limit of duration.
`Data were censored as of last visit entered into the clinical database.
`
`time of the analysis was 6, 2, and 0, respectively. Response and dura-
`tion of response data are summarized in Table 4. In the 17 previously
`treated patients with MDSs and CMML, the overall response rate was
`35%,withoutincludingpatientswhoonlyachievedmCR;ifthosepatients
`were included the response rate would be 65%. In the 15 patients with
`MDSs and CMML receiving first-line treatment, the overall response rate
`was 73% and in this group no patients achieved mCR only. Longest
`duration of response to date was 483 days overall. In one patient who
`achievedaCR,theresponsebeganbeforeoraldosingandendedincycle2,
`thus was likely attributable to the single cycle of SC azacitidine.
`No responses were observed in patients with AML. Two patients
`with AML (25%) had stable disease for 14 and 15 cycles, respectively,
`and five patients with AML (63%) received ⱖ 4 oral azacitidine cycles.
`
`DISCUSSION
`
`An oral azacitidine formulation may bring advantages for patients
`(ease of administration), society (health care cost implications), and
`disease treatment (extended administration), provided that clinical
`activity and safety are similar to SC/intravenous azacitidine. This
`phase I trial demonstrated that oral azacitidine is associated with
`minimal adverse effects at doses lower than 600 mg. The MTD was 480
`mg on a 7-day of 28 days treatment schedule. The 600 mg dose was
`associated with early onset of severe diarrhea in two of three patients.
`Diarrhea in patients taking oral azacitidine doses lower than 600 mg
`was self-limiting and manageable by treatment and/or prophylaxis
`with antidiarrheal agents and/or dose reduction. Azacitidine, along
`with one or more ingredients used in its formulation, may contribute
`to the diarrhea observed because it was a common adverse event at
`most dose levels tested. Gastrointestinal disturbances may have been
`exacerbated by the requirement to ingest oral azacitidine in a fasting
`state. Whether oral azacitidine administration with food can reduce gas-
`
`trointestinal toxicity will be evaluated in ongoing studies. Grade 3 and 4
`AEs consisted primarily of febrile neutropenia, gastrointestinal distur-
`bances, and fatigue. Of the eight patients who experienced grade 3 febrile
`neutropenia, four entered the study with a baseline ANC of ⱕ 500/␮L.
`After oral administration, maximum azacitidine plasma concen-
`trations were achieved rapidly (within 1 hour), suggesting that absorp-
`tion occurs from the proximal gastrointestinal tract. Azacitidine
`exposure increased with increasing oral doses, and the mean relative oral
`bioavailability ranged from 6.3% to 20%. After multiple doses, there was
`no evidence of azacitidine accumulation, and no apparent decline in
`absorption was seen between days 1 and 7. Azacitidine clearance was
`hepatic and extrahepatic, with little evidence of renal clearance.
`Kinetics of the change in DNA methylation levels after SC and
`oral azacitidine were similar, with maximum hypomethylation
`achieved on day 15, and methylation levels returned to near-baseline
`values by the end of each cycle. This pattern has been observed in other
`azacitidine studies.3,12 At the dosing schedule employed in this study,
`oral azacitidine affected fewer loci than SC azacitidine; however, 1,482
`loci were identified as commonly hypomethylated by both azaciti-
`dine formulations.
`Significant responses were observed in patients with MDSs and
`CMML, indicating that oral azacitidine has clinical activity in these
`settings. Although all patients received an initial cycle of SC azaciti-
`dine, which may have contributed to the clinical activity observed, it
`has been reported that only half of the total hematologic responses to
`SC azacitidine manifest within 2 cycles.13 Continued treatment with
`oral azacitidine following the single cycle of SC azacitidine is therefore
`likely to be associated with the development and/or maintenance of
`clinical responses observed in this study.
`Results from a study investigating alternative SC azacitidine dos-
`ing schedules in lower-risk patients with MDSs suggested that for all
`
`2526
`
`© 2011 by American Society of Clinical Oncology
`
`JOURNAL OF CLINICAL ONCOLOGY
`
`Downloaded from ascopubs.org by 72.165.200.210 on March 25, 2021 from 072.165.200.210
`Copyright © 2021 American Society of Clinical Oncology. All rights reserved.
`
`

`

`Phase I Study of Oral AZA in MDSs, CMML, and AML
`
`dosing regimens tested, continued azacitidine treatment may be ben-
`eficial.14 The short plasma half-life of azacitidine, S-phase restricted
`incorporation into DNA, and rapid remethylation of DNA, are con-
`tributing factors to the importance of chronic exposure to the drug. It
`is therefore likely that extended schedules of oral administration will
`positively affect clinical activity of azacitidine. A follow-up trial has
`been initiated to investigate the efficacy of such extended schedules.
`In conclusion, the MTD for oral azacitidine administered daily
`for 7 days of a 28-day cycle was determined to be 480 mg, and oral
`azacitidine is bioavailable and biologically active. Clinical responses
`were reported in 35% of previously treated patients with MDSs and
`CMML, and in 73% of patients who received oral azacitidine as first-
`line therapy. Lower drug exposure

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket